Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 9(95): eabq1558, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701190

ABSTRACT

Steroid resistance poses a major challenge for the management of autoimmune neuroinflammation. T helper 17 (TH17) cells are widely implicated in the pathology of steroid resistance; however, the underlying mechanisms are unknown. In this study, we identified that interleukin-1 receptor (IL-1R) blockade rendered experimental autoimmune encephalomyelitis (EAE) mice sensitive to dexamethasone (Dex) treatment. Interleukin-1ß (IL-1ß) induced a signal transducer and activator of transcription 5 (STAT5)-mediated steroid-resistant transcriptional program in TH17 cells, which promoted inflammatory cytokine production and suppressed Dex-induced anti-inflammatory genes. TH17-specific deletion of STAT5 ablated the IL-1ß-induced steroid-resistant transcriptional program and rendered EAE mice sensitive to Dex treatment. IL-1ß synergized with Dex to promote the STAT5-dependent expression of CD69 and the development of central nervous system (CNS)-resident CD69+ TH17 cells. Combined IL-1R blockade and Dex treatment ablated CNS-resident TH17 cells, reduced EAE severity, and prevented relapse. CD69+ tissue-resident TH17 cells were also detected in brain lesions of patients with multiple sclerosis. These findings (i) demonstrate that IL-1ß-STAT5 signaling in TH17 cells mediates steroid resistance and (ii) identify a therapeutic strategy for reversing steroid resistance in TH17-mediated CNS autoimmunity.


Subject(s)
Dexamethasone , Encephalomyelitis, Autoimmune, Experimental , Interleukin-1beta , STAT5 Transcription Factor , Th17 Cells , Animals , Th17 Cells/immunology , STAT5 Transcription Factor/metabolism , STAT5 Transcription Factor/immunology , Mice , Interleukin-1beta/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Mice, Inbred C57BL , Drug Resistance , Signal Transduction/immunology , Mice, Knockout , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/drug therapy , Female , Humans
3.
JCI Insight ; 8(18)2023 09 22.
Article in English | MEDLINE | ID: mdl-37607012

ABSTRACT

Tumor necrosis factor receptor-associated factor 4 (TRAF4) is an important regulator of type 2 responses in the airway; however, the underlying cellular and molecular mechanisms remain elusive. Herein, we generated T cell-specific TRAF4-deficient (CD4-cre Traf4fl/fl) mice and investigated the role of TRAF4 in memory Th2 cells expressing IL-33 receptor (ST2, suppression of tumorigenicity 2) (ST2+ mTh2 cells) in IL-33-mediated type 2 airway inflammation. We found that in vitro-polarized TRAF4-deficient (CD4-cre Traf4fl/fl) ST2+ mTh2 cells exhibited decreased IL-33-induced proliferation as compared with TRAF4-sufficient (Traf4fl/fl) cells. Moreover, CD4-cre Traf4fl/fl mice showed less ST2+ mTh2 cell proliferation and eosinophilic infiltration in the lungs than Traf4fl/fl mice in the preclinical models of IL-33-mediated type 2 airway inflammation. Mechanistically, we discovered that TRAF4 was required for the activation of AKT/mTOR and ERK1/2 signaling pathways as well as the expression of transcription factor Myc and nutrient transporters (Slc2a1, Slc7a1, and Slc7a5), signature genes involved in T cell growth and proliferation, in ST2+ mTh2 cells stimulated by IL-33. Taken together, the current study reveals a role of TRAF4 in ST2+ mTh2 cells in IL-33-mediated type 2 pulmonary inflammation, opening up avenues for the development of new therapeutic strategies.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Animals , Mice , Cell Proliferation , Inflammation/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-33/genetics , Interleukin-33/metabolism , Lung/metabolism , Th2 Cells/metabolism , TNF Receptor-Associated Factor 4/metabolism
4.
Hepatol Commun ; 7(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-37185170

ABSTRACT

BACKGROUND: Macrophage-inducible C-type lectin (Mincle) is expressed on hepatic macrophages and senses ethanol (EtOH)-induced danger signals released from dying hepatocytes and promotes IL-1ß production. However, it remains unclear what and how EtOH-induced Mincle ligands activate downstream signaling events to mediate IL-1ß release and contribute to alcohol-associated liver disease (ALD). In this study, we investigated the association of circulating ß-glucosylceramide (ß-GluCer), an endogenous Mincle ligand, with severity of ALD and examined the mechanism by which ß-GluCer engages Mincle on hepatic macrophages to release IL-1ß in the absence of cell death and exacerbates ALD. METHOD AND RESULTS: Concentrations of ß-GluCer were increased in serum of patients with severe AH and correlated with disease severity. Challenge of hepatic macrophages with lipopolysaccharide and ß-GluCer induced formation of a Mincle and Gsdmd-dependent secretory complex containing chaperoned full-length gasdermin D (Hsp90-CDC37-NEDD4) with polyubiquitinated pro-IL-1ß and components of the Caspase 8-NLRP3 inflammasome loaded as cargo in small extracellular vesicles (sEVs). Gao-binge EtOH exposure to wild-type, but not Mincle-/- and Gsdmd-/-, mice increased release of IL-1ß-containing sEVs from liver explant cultures. Myeloid-specific deletion of Gsdmd similarly decreased the formation of sEVs by liver explant cultures and protected mice from EtOH-induced liver injury. sEVs collected from EtOH-fed wild-type, but not Gsdmd-/-, mice promoted injury of cultured hepatocytes and, when injected into wild-type mice, aggravated Gao-binge EtOH-induced liver injury. CONCLUSION: ß-GluCer functions as a danger-associated molecular pattern activating Mincle-dependent gasdermin D-mediated formation and release of IL-1ß-containing sEVs, which in turn exacerbate hepatocyte cell death and contribute to the pathogenesis of ALD.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Liver Diseases, Alcoholic , Animals , Mice , Ethanol/toxicity , Gasdermins , Kupffer Cells/metabolism , Liver Diseases, Alcoholic/metabolism
5.
bioRxiv ; 2023 Feb 12.
Article in English | MEDLINE | ID: mdl-36798382

ABSTRACT

Tumor necrosis factor receptor (TNF)-associated factor 4 (TRAF4) is an important regulator of type 2 responses in the airway; however, the underlying cellular and molecular mechanisms remain elusive. Herein, we generated T cell-specific TRAF4-deficient (CD4cre-Traf4fl/fl) mice and investigated the role of TRAF4 in interleukin (IL)-33 receptor (ST2, suppression of tumorigenicity 2)-expressing memory Th2 cells (ST2+ mTh2) in IL-33-mediated type 2 airway inflammation. We found that in vitro polarized TRAF4-deficient (CD4cre- Traf4fl/fl) ST2+ mTh2 cells exhibited decreased IL-33-induced proliferation as compared with TRAF4-sufficient (Traf4fl/fl) cells. Moreover, CD4cre-Traf4fl/fl mice showed less ST2+ mTh2 cell proliferation and eosinophilic infiltration in the lungs than Traf4fl/fl mice in the preclinical models of IL-33-mediated type 2 airway inflammation. Mechanistically, we discovered that TRAF4 was required for the activation of AKT/mTOR and ERK1/2 signaling pathways as well as the expression of transcription factor Myc and nutrient transporters (Slc2a1, Slc7a1, and Slc7a5), signature genes involved in T cell growth and proliferation, in ST2+ mTh2 cells stimulated by IL-33. Taken together, the current study reveals a previously unappreciated role of TRAF4 in ST2+ mTh2 cells in IL-33-mediated type 2 pulmonary inflammation, opening up avenues for the development of new therapeutic strategies.

6.
EMBO J ; 42(1): e110780, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36373462

ABSTRACT

IL-1ß can exit the cytosol as an exosomal cargo following inflammasome activation in intestinal epithelial cells (IECs) in a Gasdermin D (GSDMD)-dependent manner. The mechanistic connection linking inflammasome activation and the biogenesis of exosomes has so far remained largely elusive. Here, we report the Ras GTPase-activating-like protein IQGAP1 functions as an adaptor, bridging GSDMD to the endosomal sorting complexes required for transport (ESCRT) machinery to promote the biogenesis of pro-IL-1ß-containing exosomes in response to NLPR3 inflammasome activation. We identified IQGAP1 as a GSDMD-interacting protein through a non-biased proteomic analysis. Functional investigation indicated the IQGAP1-GSDMD interaction is required for LPS and ATP-induced exosome release. Further analysis revealed that IQGAP1 serves as an adaptor which bridges GSDMD and associated IL-1ß complex to Tsg101, a component of the ESCRT complex, and enables the packaging of GSDMD and IL-1ß into exosomes. Importantly, this process is dependent on an LPS-induced increase in GTP-bound CDC42, a small GTPase known to activate IQGAP1. Taken together, this study reveals IQGAP1 as a link between inflammasome activation and GSDMD-dependent, ESCRT-mediated exosomal release of IL-1ß.


Subject(s)
Exosomes , Inflammasomes , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Gasdermins , Exosomes/metabolism , ras Proteins/metabolism , Lipopolysaccharides/pharmacology , Proteomics , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Interleukin-1beta/metabolism , Pyroptosis
7.
J Immunol ; 209(10): 1860-1869, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36426949

ABSTRACT

IL-17A plays an important role in the pathogenesis of asthma, particularly the neutrophilic corticosteroid (CS)-resistant subtype of asthma. Clinical studies suggest that a subset of asthma patients, i.e., Th17/IL-17A-mediated (type 17) CS-resistant neutrophilic asthma, may improve with Th17/IL-17A pathway blockade. However, little is known about the mechanisms underlying type 17 asthma and CS response. In this article, we show that blood levels of lipocalin-2 (LCN2) and serum amyloid A (SAA) levels are positively correlated with IL-17A levels and are not inhibited by high-dose CS usage in asthma patients. In airway cell culture systems, IL-17A induces these two secreted proteins, and their induction is enhanced by CS. Furthermore, plasma LCN2 and SAA levels are increased in mice on a preclinical type 17 asthma model, correlated to IL-17A levels, and are not reduced by glucocorticoid (GC). In the mechanistic studies, we identify CEBPB as the critical transcription factor responsible for the synergistic induction of LCN2 and SAA by IL-17A and GC. IL-17A and GC collaboratively regulate CEBPB at both transcriptional and posttranscriptional levels. The posttranscriptional regulation of CEBPB is mediated in part by Act1, the adaptor and RNA binding protein in IL-17A signaling, which directly binds CEBPB mRNA and inhibits its degradation. Overall, our findings suggest that blood LCN2 and SAA levels may be associated with a type 17 asthma subtype and provide insight into the molecular mechanism of the IL-17A-Act1/CEBPB axis on these CS-resistant genes.


Subject(s)
Asthma , Interleukin-17 , Mice , Animals , Interleukin-17/genetics , Asthma/drug therapy , Asthma/pathology , Th17 Cells/pathology , Signal Transduction , Glucocorticoids
8.
J Interferon Cytokine Res ; 42(9): 482-492, 2022 09.
Article in English | MEDLINE | ID: mdl-35900274

ABSTRACT

Colorectal carcinoma is the leading cause of cancer-related death. Previously we have shown that tumor suppressor single immunoglobulin interleukin-1-related receptor (SIGIRR) is frequently inactivated in human colorectal cancer by the increased expression of a novel SIGIRR isoform (SIGIRRΔE8). SIGIRRΔE8 showed increased retention in the cytoplasm and loss of complex glycan modification compared to the full-length SIGIRR. Now we found that the arginine residues located in the C-terminus of SIGIRRΔE8 serve as an endoplasmic reticulum retention signal and are required for resident protein ribophorin 1 (RPN1) interaction. In addition, we found that SIGIRRΔE8 exerts a direct impact on cell metabolism through interaction with the adenosine triphosphate synthase in the colorectal cancer cells. SIGIRRΔE8 expression promoted the metabolic shift through upregulation of mammalian target of rapamycin signaling pathway and dysregulation of mitochondrial function to promote survival and proliferation of colon cancer cells in xenograft model.


Subject(s)
Colonic Neoplasms , Receptors, Interleukin-1/metabolism , Adenosine Triphosphate/metabolism , Arginine/metabolism , Colonic Neoplasms/pathology , Humans , Immunoglobulins/metabolism , Interleukin-1 , Metabolic Networks and Pathways , TOR Serine-Threonine Kinases/metabolism
9.
Nat Commun ; 13(1): 2406, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35504893

ABSTRACT

The C-type lectin receptor Mincle is known for its important role in innate immune cells in recognizing pathogen and damage associated molecular patterns. Here we report a T cell-intrinsic role for Mincle in the pathogenesis of experimental autoimmune encephalomyelitis (EAE). Genomic deletion of Mincle in T cells impairs TH17, but not TH1 cell-mediated EAE, in alignment with significantly higher expression of Mincle in TH17 cells than in TH1 cells. Mechanistically, dying cells release ß-glucosylceramide during inflammation, which serves as natural ligand for Mincle. Ligand engagement induces activation of the ASC-NLRP3 inflammasome, which leads to Caspase8-dependent IL-1ß production and consequentially TH17 cell proliferation via an autocrine regulatory loop. Chemical inhibition of ß-glucosylceramide synthesis greatly reduces inflammatory CD4+ T cells in the central nervous system and inhibits EAE progression in mice. Taken together, this study indicates that sensing of danger signals by Mincle on TH17 cells plays a critical role in promoting CNS inflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Th17 Cells , Animals , Central Nervous System/metabolism , Glucosylceramides/metabolism , Inflammation/metabolism , Ligands , Mice
10.
Cell ; 185(2): 283-298.e17, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35021065

ABSTRACT

Gasdermins are a family of structurally related proteins originally described for their role in pyroptosis. Gasdermin B (GSDMB) is currently the least studied, and while its association with genetic susceptibility to chronic mucosal inflammatory disorders is well established, little is known about its functional relevance during active disease states. Herein, we report increased GSDMB in inflammatory bowel disease, with single-cell analysis identifying epithelial specificity to inflamed colonocytes/crypt top colonocytes. Surprisingly, mechanistic experiments and transcriptome profiling reveal lack of inherent GSDMB-dependent pyroptosis in activated epithelial cells and organoids but instead point to increased proliferation and migration during in vitro wound closure, which arrests in GSDMB-deficient cells that display hyper-adhesiveness and enhanced formation of vinculin-based focal adhesions dependent on PDGF-A-mediated FAK phosphorylation. Importantly, carriage of disease-associated GSDMB SNPs confers functional defects, disrupting epithelial restitution/repair, which, altogether, establishes GSDMB as a critical factor for restoration of epithelial barrier function and the resolution of inflammation.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis , Base Sequence , Case-Control Studies , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial Cells/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HEK293 Cells , HT29 Cells , Humans , Inflammatory Bowel Diseases/genetics , Methotrexate/pharmacology , Mutation/genetics , Phosphorylation/drug effects , Polymorphism, Single Nucleotide/genetics , Pyroptosis/drug effects , Pyroptosis/genetics , Reproducibility of Results , Transcriptome/drug effects , Transcriptome/genetics , Up-Regulation/drug effects , Wound Healing/drug effects , Wound Healing/genetics
12.
J Clin Invest ; 130(8): 4218-4234, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32597834

ABSTRACT

Gasdermin D (GSDMD) induces pyroptosis via the pore-forming activity of its N-terminal domain, cleaved by activated caspases associated with the release of IL-1ß. Here, we report a nonpyroptotic role of full-length GSDMD in guiding the release of IL-1ß-containing small extracellular vesicles (sEVs) from intestinal epithelial cells (IECs). In response to caspase-8 inflammasome activation, GSDMD, chaperoned by Cdc37/Hsp90, recruits the E3 ligase, NEDD4, to catalyze polyubiquitination of pro-IL-1ß, serving as a signal for cargo loading into secretory vesicles. GSDMD and IL-1ß colocalize with the exosome markers CD63 and ALIX intracellularly, and GSDMD and NEDD4 are required for release of CD63+ sEVs containing IL-1ß, GSDMD, NEDD4, and caspase-8. Importantly, increased expression of epithelial-derived GSDMD is observed both in patients with inflammatory bowel disease (IBD) and those with experimental colitis. While GSDMD-dependent release of IL-1ß-containing sEVs is detected in cultured colonic explants from colitic mice, GSDMD deficiency substantially attenuates disease severity, implicating GSDMD-mediated release of IL-1ß sEVs in the pathogenesis of intestinal inflammation, such as that observed in IBD.


Subject(s)
Colitis/metabolism , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/metabolism , Interleukin-1beta/metabolism , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Animals , Cell Line , Colitis/genetics , Colitis/pathology , Epithelial Cells/pathology , Exosomes/genetics , Exosomes/metabolism , Exosomes/pathology , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Interleukin-1beta/genetics , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Phosphate-Binding Proteins/genetics , Tetraspanin 30/genetics , Tetraspanin 30/metabolism
13.
Nat Commun ; 11(1): 900, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32060280

ABSTRACT

Copper levels are known to be elevated in inflamed and malignant tissues. But the mechanism underlying this selective enrichment has been elusive. In this study, we report a axis by which inflammatory cytokines, such as IL-17, drive cellular copper uptake via the induction of a metalloreductase, STEAP4. IL-17-induced elevated intracellular copper level leads to the activation of an E3-ligase, XIAP, which potentiates IL-17-induced NFκB activation and suppresses the caspase 3 activity. Importantly, this IL-17-induced STEAP4-dependent cellular copper uptake is critical for colon tumor formation in a murine model of colitis-associated tumorigenesis and STEAP4 expression correlates with IL-17 level and XIAP activation in human colon cancer. In summary, this study reveals a IL-17-STEAP4-XIAP axis through which the inflammatory response induces copper uptake, promoting colon tumorigenesis.


Subject(s)
Colon/immunology , Colonic Neoplasms/immunology , Copper/metabolism , Inhibitor of Apoptosis Proteins/immunology , Interleukin-17/immunology , Membrane Proteins/immunology , Animals , Carcinogenesis , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colon/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Copper/immunology , Humans , Inhibitor of Apoptosis Proteins/genetics , Interleukin-17/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
Elife ; 92020 01 28.
Article in English | MEDLINE | ID: mdl-31989925

ABSTRACT

Lung disease causes significant morbidity and mortality, and is exacerbated by environmental injury, for example through lipopolysaccharide (LPS) or ozone (O3). Toll-like receptors (TLRs) orchestrate immune responses to injury by recognizing pathogen- or danger-associated molecular patterns. TLR4, the prototypic receptor for LPS, also mediates inflammation after O3, triggered by endogenous hyaluronan. Regulation of TLR4 signaling is incompletely understood. TLR5, the flagellin receptor, is expressed in alveolar macrophages, and regulates immune responses to environmental injury. Using in vivo animal models of TLR4-mediated inflammations (LPS, O3, hyaluronan), we show that TLR5 impacts the in vivo response to LPS, hyaluronan and O3. We demonstrate that immune cells of human carriers of a dominant negative TLR5 allele have decreased inflammatory response to O3 exposure ex vivo and LPS exposure in vitro. Using primary murine macrophages, we find that TLR5 physically associates with TLR4 and biases TLR4 signaling towards the MyD88 pathway. Our results suggest an updated paradigm for TLR4/TLR5 signaling.


Immune cells in the lung help guard against infections. On the surface of these cells are proteins called TLR receptors that recognize dangerous molecules or DNA from disease-causing microbes such as bacteria. When the immune cells detect these invaders, the TLR receptors spring into action and trigger an inflammatory response to destroy the microbes. This inflammation usually helps the lung clear infections. But it can also be harmful and damage the lung, for example when inflammation is caused by non-infectious substances such as pollutants in the atmosphere. There are several TLR receptors that each recognize a specific molecule. In 2010, researchers showed that the receptor TLR4 is responsible for causing inflammation in the lung after exposure to pollution. Another receptor called TLR5 also helps activate the immune response in the lung. But it was unclear whether this receptor also plays a role in pollution-linked lung damage. Now, Hussain, Johnson, Sciurba et al. ­ including one of the researchers involved in the 2010 study ­ have investigated the role of TLR5 in immune cells from the lungs of humans and mice. The experiments showed that TLR5 works together with TLR4 and helps trigger an inflammatory response to both pollutants and bacteria. Hussain et al. found that people lacking a working TLR5 receptor (which make up 3­10% of the population) are less likely to experience lung inflammation when exposed to pollution or bacterial proteins that activate TLR4. These findings suggest that people without TLR5 may be protected from pollution-induced lung injury. Further research into the role of TLR5 could help develop genetic tests for identifying people who are more sensitive to damage from pollution. This information could then be used to determine the likelihood of a patient experiencing certain lung diseases.


Subject(s)
Lung Injury , Myeloid Differentiation Factor 88 , Signal Transduction , Toll-Like Receptor 4 , Toll-Like Receptor 5 , Animals , Cells, Cultured , Humans , Inflammation/immunology , Inflammation/metabolism , Lung/immunology , Lung/metabolism , Lung Injury/chemically induced , Lung Injury/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Polymorphism, Single Nucleotide/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism
15.
Front Immunol ; 10: 1293, 2019.
Article in English | MEDLINE | ID: mdl-31316496

ABSTRACT

Tumor necrosis factor receptor (TNFR)-associated factors or (TRAFs) are important mediators of Interleukin-17 (IL-17) cytokine signaling and contribute to driving tissue responses that are crucial for protective immunity but are often implicated in immunopathology. By amplifying tissue immune activity, IL-17 cytokine pathways contribute to maintaining barrier function as well as activation of innate and adaptive immunity necessary for host defense. IL-17 receptors signaling is orchestrated in part, by the engagement of TRAFs and the subsequent unlocking of downstream cellular machinery that can promote pathogen clearance or contribute to immune dysregulation, chronic inflammation, and disease. Originally identified as signaling adaptors for TNFR superfamily, TRAF proteins can mediate the signaling of a variety of intercellular and extracellular stimuli and have been shown to regulate the downstream activity of many cytokine receptors including receptors for IL-1ß, IL-2, IL-6, IL-17, IL-18, IL-33, type I IFNs, type III IFNs, GM-CSF, M-CSF, and TGF-ß Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I- like receptors, and C-type lectin receptors. This review will focus on discussing studies that reveal our current understanding of how TRAFs mediate and regulate biochemical activities downstream of the IL-17 cytokines signaling.


Subject(s)
Cytokines/immunology , Interleukin-17/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/immunology , Adaptive Immunity/immunology , Animals , Cytokines/metabolism , Humans , Immunity, Innate/immunology , Interleukin-17/metabolism , NLR Proteins/immunology , NLR Proteins/metabolism , Receptors, Cytokine/immunology , Receptors, Cytokine/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism
16.
J Immunol ; 202(5): 1540-1548, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30683702

ABSTRACT

IL-17A is a critical proinflammatory cytokine for the pathogenesis of asthma including neutrophilic pulmonary inflammation and airway hyperresponsiveness. In this study, by cell type-specific deletion of IL-17R and adaptor Act1, we demonstrated that IL-17R/Act1 exerts a direct impact on the contraction of airway smooth muscle cells (ASMCs). Mechanistically, IL-17A induced the recruitment of Rab35 (a small monomeric GTPase) and DennD1C (guanine nucleotide exchange factor [GEF]) to the IL-17R/Act1 complex in ASMCs, resulting in activation of Rab35. Rab35 knockdown showed that IL-17A-induced Rab35 activation was essential for protein kinase Cα (PKCα) activation and phosphorylation of fascin at Ser39 in ASMCs, allowing F-actin to interact with myosin to form stress fibers and enhance the contraction induced by methacholine. PKCα inhibitor or Rab35 knockdown indeed substantially reduced IL-17A-induced stress fiber formation in ASMCs and attenuated IL-17A-enhanced, methacholine-induced contraction of airway smooth muscle. Taken together, these data indicate that IL-17A promotes airway smooth muscle contraction via direct recruitment of Rab35 to IL-17R, followed by PKCα activation and stress fiber formation.


Subject(s)
Interleukin-17/metabolism , Muscle, Smooth/metabolism , Protein Kinase C-alpha/antagonists & inhibitors , Receptors, Interleukin-17/metabolism , Stress Fibers/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Interleukin-17/antagonists & inhibitors , Interleukin-17/deficiency , Mice , Mice, Knockout , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Interleukin-17/antagonists & inhibitors , Stress Fibers/drug effects , rab GTP-Binding Proteins/antagonists & inhibitors
17.
Nat Immunol ; 19(4): 354-365, 2018 04.
Article in English | MEDLINE | ID: mdl-29563620

ABSTRACT

Mechanisms that degrade inflammatory mRNAs are well known; however, stabilizing mechanisms are poorly understood. Here, we show that Act1, an interleukin-17 (IL-17)-receptor-complex adaptor, binds and stabilizes mRNAs encoding key inflammatory proteins. The Act1 SEFIR domain binds a stem-loop structure, the SEFIR-binding element (SBE), in the 3' untranslated region (UTR) of Cxcl1 mRNA, encoding an inflammatory chemokine. mRNA-bound Act1 directs formation of three compartmentally distinct RNA-protein complexes (RNPs) that regulate three disparate events in inflammatory-mRNA metabolism: preventing mRNA decay in the nucleus, inhibiting mRNA decapping in P bodies and promoting translation. SBE RNA aptamers decreased IL-17-mediated mRNA stabilization in vitro, IL-17-induced skin inflammation and airway inflammation in a mouse asthma model, thus providing a therapeutic strategy for autoimmune diseases. These results reveal a network in which Act1 assembles RNPs on the 3' UTRs of select mRNAs and consequently controls receptor-mediated mRNA stabilization and translation during inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Inflammation/immunology , Interleukin-17/metabolism , RNA Stability/physiology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , Gene Expression Regulation/immunology , Inflammation/metabolism , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Receptors, Interleukin-17/metabolism
18.
Elife ; 62017 10 09.
Article in English | MEDLINE | ID: mdl-28990926

ABSTRACT

Expression of inflammatory genes is determined in part by post-transcriptional regulation of mRNA metabolism but how stimulus- and transcript-dependent nuclear export influence is poorly understood. Here, we report a novel pathway in which LPS/TLR4 engagement promotes nuclear localization of IRAK2 to facilitate nuclear export of a specific subset of inflammation-related mRNAs for translation in murine macrophages. IRAK2 kinase activity is required for LPS-induced RanBP2-mediated IRAK2 sumoylation and subsequent nuclear translocation. Array analysis showed that an SRSF1-binding motif is enriched in mRNAs dependent on IRAK2 for nuclear export. Nuclear IRAK2 phosphorylates SRSF1 to reduce its binding to target mRNAs, which promotes the RNA binding of the nuclear export adaptor ALYREF and nuclear export receptor Nxf1 loading for the export of the mRNAs. In summary, LPS activates a nuclear function of IRAK2 that facilitates the assembly of nuclear export machinery to export selected inflammatory mRNAs to the cytoplasm for translation.


Subject(s)
Active Transport, Cell Nucleus , Interleukin-1 Receptor-Associated Kinases/metabolism , Macrophages/immunology , RNA, Messenger/metabolism , Animals , Lipopolysaccharides/metabolism , Macrophages/drug effects , Mice , Nucleocytoplasmic Transport Proteins/metabolism , Phosphorylation , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors/metabolism , Sumoylation
19.
Nat Commun ; 8: 15508, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28561022

ABSTRACT

NOTCH1 signalling contributes to defective remyelination by impairing differentiation of oligodendrocyte progenitor cells (OPCs). Here we report that IL-17 stimulation induces NOTCH1 activation in OPCs, contributing to Th17-mediated demyelinating disease. Mechanistically, IL-17R interacts with NOTCH1 via the extracellular domain, which facilitates the cleavage of NOTHC1 intracellular domain (NICD1). IL-17-induced NOTCH1 activation results in the interaction of IL-17R adaptor Act1 with NICD1, followed by the translocation of the Act1-NICD1 complex into the nucleus. Act1-NICD1 are recruited to the promoters of several NOTCH1 target genes (including STEAP4, a metalloreductase important for inflammation and cell proliferation) that are specifically induced in the spinal cord by Th17 cells. A decoy peptide disrupting the IL-17RA-NOTCH1 interaction inhibits IL-17-induced NOTCH1 activation and attenuates Th17-mediated experimental autoimmune encephalitis (EAE). Taken together, these findings demonstrate critical crosstalk between the IL-17 and NOTCH1 pathway, regulating Th17-induced inflammatory and proliferative genes to promote demyelinating disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/metabolism , Multiple Sclerosis/immunology , Oligodendrocyte Precursor Cells/physiology , Receptor, Notch1/immunology , Th17 Cells/immunology , Adaptor Proteins, Signal Transducing , Animals , Astrocytes , Cell Differentiation/immunology , Cell Proliferation/physiology , Coculture Techniques , Female , HEK293 Cells , HeLa Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/immunology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Protein Binding/immunology , Protein Domains/physiology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Interleukin-17/metabolism , Remyelination/physiology , Signal Transduction/immunology , Th1 Cells/immunology , Th17 Cells/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism
20.
Cell ; 168(5): 789-800.e10, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235196

ABSTRACT

The molecular basis of the incomplete penetrance of monogenic disorders is unclear. We describe here eight related individuals with autosomal recessive TIRAP deficiency. Life-threatening staphylococcal disease occurred during childhood in the proband, but not in the other seven homozygotes. Responses to all Toll-like receptor 1/2 (TLR1/2), TLR2/6, and TLR4 agonists were impaired in the fibroblasts and leukocytes of all TIRAP-deficient individuals. However, the whole-blood response to the TLR2/6 agonist staphylococcal lipoteichoic acid (LTA) was abolished only in the index case individual, the only family member lacking LTA-specific antibodies (Abs). This defective response was reversed in the patient, but not in interleukin-1 receptor-associated kinase 4 (IRAK-4)-deficient individuals, by anti-LTA monoclonal antibody (mAb). Anti-LTA mAb also rescued the macrophage response in mice lacking TIRAP, but not TLR2 or MyD88. Thus, acquired anti-LTA Abs rescue TLR2-dependent immunity to staphylococcal LTA in individuals with inherited TIRAP deficiency, accounting for incomplete penetrance. Combined TIRAP and anti-LTA Ab deficiencies underlie staphylococcal disease in this patient.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Lipopolysaccharides/metabolism , Membrane Glycoproteins/deficiency , Receptors, Interleukin-1/deficiency , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Teichoic Acids/metabolism , Adaptive Immunity , Child , Female , Fibroblasts/metabolism , Humans , Immunity, Innate , Lipopolysaccharides/immunology , Macrophages/immunology , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Monocytes/metabolism , Myeloid Differentiation Factor 88/metabolism , Pedigree , Phagocytes/metabolism , Point Mutation , Protein Isoforms/analysis , Protein Isoforms/genetics , Receptors, Interleukin-1/analysis , Receptors, Interleukin-1/genetics , Staphylococcal Infections/drug therapy , Teichoic Acids/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...